Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 19(9): e1011446, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37733807

RESUMEN

Zika virus (ZIKV) is a Flavivirus responsible for recent epidemics in Pacific Islands and in the Americas. In humans, the consequences of ZIKV infection range from asymptomatic infection to severe neurological disease such as Guillain-Barré syndrome or fetal neurodevelopmental defects, suggesting, among other factors, the influence of host genetic variants. We previously reported similar diverse outcomes of ZIKV infection in mice of the Collaborative Cross (CC), a collection of inbred strains with large genetic diversity. CC071/TauUnc (CC071) was the most susceptible CC strain with severe symptoms and lethality. Notably, CC071 has been recently reported to be also susceptible to other flaviviruses including dengue virus, Powassan virus, West Nile virus, and to Rift Valley fever virus. To identify the genetic origin of this broad susceptibility, we investigated ZIKV replication in mouse embryonic fibroblasts (MEFs) from CC071 and two resistant strains. CC071 showed uncontrolled ZIKV replication associated with delayed induction of type-I interferons (IFN-I). Genetic analysis identified a mutation in the Irf3 gene specific to the CC071 strain which prevents the protein phosphorylation required to activate interferon beta transcription. We demonstrated that this mutation induces the same defective IFN-I response and uncontrolled viral replication in MEFs as an Irf3 knock-out allele. By contrast, we also showed that Irf3 deficiency did not induce the high plasma viral load and clinical severity observed in CC071 mice and that susceptibility alleles at other genes, not associated with the IFN-I response, are required. Our results provide new insight into the in vitro and in vivo roles of Irf3, and into the genetic complexity of host responses to flaviviruses.


Asunto(s)
Flavivirus , Interferón Tipo I , Infección por el Virus Zika , Virus Zika , Animales , Ratones , Ratones de Colaboración Cruzada , Fibroblastos , Factor 3 Regulador del Interferón/genética , Virus Zika/genética , Infección por el Virus Zika/genética
2.
EMBO Rep ; 24(4): e56055, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-36876574

RESUMEN

Bat sarbecovirus BANAL-236 is highly related to SARS-CoV-2 and infects human cells, albeit lacking the furin cleavage site in its spike protein. BANAL-236 replicates efficiently and pauci-symptomatically in humanized mice and in macaques, where its tropism is enteric, strongly differing from that of SARS-CoV-2. BANAL-236 infection leads to protection against superinfection by a virulent strain. We find no evidence of antibodies recognizing bat sarbecoviruses in populations in close contact with bats in which the virus was identified, indicating that such spillover infections, if they occur, are rare. Six passages in humanized mice or in human intestinal cells, mimicking putative early spillover events, select adaptive mutations without appearance of a furin cleavage site and no change in virulence. Therefore, acquisition of a furin site in the spike protein is likely a pre-spillover event that did not occur upon replication of a SARS-CoV-2-like bat virus in humans or other animals. Other hypotheses regarding the origin of the SARS-CoV-2 should therefore be evaluated, including the presence of sarbecoviruses carrying a spike with a furin cleavage site in bats.


Asunto(s)
COVID-19 , Humanos , Animales , Ratones , SARS-CoV-2 , Furina/genética , Furina/metabolismo , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Mutación
3.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33649222

RESUMEN

Natural killer (NK) cells are innate effectors armed with cytotoxic and cytokine-secreting capacities whose spontaneous antitumor activity is key to numerous immunotherapeutic strategies. However, current mouse models fail to mirror the extensive immune system variation that exists in the human population which may impact on NK cell-based therapies. We performed a comprehensive profiling of NK cells in the Collaborative Cross (CC), a collection of novel recombinant inbred mouse strains whose genetic diversity matches that of humans, thereby providing a unique and highly diverse small animal model for the study of immune variation. We demonstrate that NK cells from CC strains displayed a breadth of phenotypic and functional variation reminiscent of that reported for humans with regards to cell numbers, key marker expression, and functional capacities. We took advantage of the vast genetic diversity of the CC and identified nine genomic loci through quantitative trait locus mapping driving these phenotypic variations. SNP haplotype patterns and variant effect analyses identified candidate genes associated with lung NK cell numbers, frequencies of CD94+ NK cells, and expression levels of NKp46. Thus, we demonstrate that the CC represents an outstanding resource to study NK cell diversity and its regulation by host genetics.


Asunto(s)
Antígenos Ly , Regulación de la Expresión Génica/inmunología , Células Asesinas Naturales/inmunología , Subfamília D de Receptores Similares a Lectina de las Células NK , Receptor 1 Gatillante de la Citotoxidad Natural , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo/inmunología , Animales , Antígenos Ly/genética , Antígenos Ly/inmunología , Cruzamientos Genéticos , Ratones , Ratones Endogámicos , Subfamília D de Receptores Similares a Lectina de las Células NK/genética , Subfamília D de Receptores Similares a Lectina de las Células NK/inmunología , Receptor 1 Gatillante de la Citotoxidad Natural/genética , Receptor 1 Gatillante de la Citotoxidad Natural/inmunología
4.
J Biol Chem ; 298(1): 101290, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34678315

RESUMEN

The current COVID-19 pandemic illustrates the importance of obtaining reliable methods for the rapid detection of SARS-CoV-2. A highly specific and sensitive diagnostic test able to differentiate the SARS-CoV-2 virus from common human coronaviruses is therefore needed. Coronavirus nucleoprotein (N) localizes to the cytoplasm and the nucleolus and is required for viral RNA synthesis. N is the most abundant coronavirus protein, so it is of utmost importance to develop specific antibodies for its detection. In this study, we developed a sandwich immunoassay to recognize the SARS-CoV-2 N protein. We immunized one alpaca with recombinant SARS-CoV-2 N and constructed a large single variable domain on heavy chain (VHH) antibody library. After phage display selection, seven VHHs recognizing the full N protein were identified by ELISA. These VHHs did not recognize the nucleoproteins of the four common human coronaviruses. Hydrogen Deuterium eXchange-Mass Spectrometry (HDX-MS) analysis also showed that these VHHs mainly targeted conformational epitopes in either the C-terminal or the N-terminal domains. All VHHs were able to recognize SARS-CoV-2 in infected cells or on infected hamster tissues. Moreover, the VHHs could detect the SARS variants B.1.17/alpha, B.1.351/beta, and P1/gamma. We propose that this sandwich immunoassay could be applied to specifically detect the SARS-CoV-2 N in human nasal swabs.


Asunto(s)
Ensayo de Inmunoadsorción Enzimática/métodos , Proteínas de la Nucleocápside/análisis , SARS-CoV-2/inmunología , Anticuerpos de Dominio Único/inmunología , Animales , Cricetinae , Electroforesis en Gel de Poliacrilamida , Humanos , Límite de Detección , Proteínas de la Nucleocápside/inmunología
5.
J Neuroinflammation ; 19(1): 307, 2022 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-36539803

RESUMEN

BACKGROUND: Zika virus (ZIKV) infection at postnatal or adult age can lead to neurological disorders associated with cognitive defects. Yet, how mature neurons respond to ZIKV remains substantially unexplored. METHODS: The impact of ZIKV infection on mature neurons and microglia was analyzed at the molecular and cellular levels, in vitro using immunocompetent primary cultured neurons and microglia, and in vivo in the brain of adult immunocompetent mice following intracranial ZIKV inoculation. We have used C57BL/6 and the genetically diverse Collaborative Cross mouse strains, displaying a broad range of susceptibility to ZIKV infection, to question the correlation between the effects induced by ZIKV infection on neurons and microglia and the in vivo susceptibility to ZIKV. RESULTS: As a result of a delayed induction of interferon beta (IFNB) expression and response, infected neurons displayed an inability to stop ZIKV replication, a trait that was further increased in neurons from susceptible mice. Alongside with an enhanced expression of ZIKV RNA, we observed in vivo, in the brain of susceptible mice, an increased level of active Iba1-expressing microglial cells occasionally engulfing neurons and displaying a gene expression profile close to the molecular signature of disease-associated microglia (DAM). In vivo as well as in vitro, only neurons and not microglial cells were identified as infected, raising the question of the mechanisms underlying microglia activation following brain ZIKV infection. Treatment of primary cultured microglia with conditioned media from ZIKV-infected neurons demonstrated that type-I interferons (IFNs-I) secreted by neurons late after infection activate non-infected microglial cells. In addition, ZIKV infection induced pathological phosphorylation of Tau (pTau) protein, a hallmark of neurodegenerative tauopathies, in vitro and in vivo with clusters of neurons displaying pTau surrounded by active microglial cells. CONCLUSIONS: We show that ZIKV-infected mature neurons display an inability to stop viral replication in link with a delayed IFNB expression and response, while signaling microglia for activation through IFNs-I secreted at late times post-infection. In the brain of ZIKV-infected susceptible mice, uninfected microglial cells adopt an active morphology and a DAM expression profile, surrounding and sometimes engulfing neurons while ZIKV-infected neurons accumulate pTau, overall reflecting a tauopathy-like phenotype.


Asunto(s)
Tauopatías , Infección por el Virus Zika , Virus Zika , Ratones , Animales , Infección por el Virus Zika/metabolismo , Virus Zika/genética , Interferón beta/genética , Ratones Endogámicos C57BL , Neuronas/metabolismo , Tauopatías/patología , Replicación Viral , Fenotipo
6.
Eur J Neurol ; 29(9): 2823-2831, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35699338

RESUMEN

BACKGROUND AND PURPOSE: Zika virus (ZIKV) infection has been associated with Guillain-Barré syndrome (GBS). However, little is known about the consequence of ZIKV infection on olfaction in humans. METHODS: Immediately before the COVID-19 outbreak, we prospectively investigated the olfactory capacities of 19 patients with ZIKV-associated GBS from the French West Indies and compared them to nine controls from the same population, with GBS of similar severity but independent of ZIKV infection. To provide further evidence that ZIKV infection induces smell alteration, we investigated the consequences of ZIKV infection on olfactory abilities using a mouse model. RESULTS: Patients with GBS-ZIKA+ had poorer olfactory function than GBS-non-ZIKA, even 1-2 years after the acute phase. The proportion of patients with hyposmia was significantly higher in the GBS-ZIKA+ than in the GBS-non-ZIKA group (68.4% vs. 22.2%, p = 0.042). These deficits were characterized by lower threshold and identification scores and were independent from GBS severity. Additionally, ZIKV infection was found to impair olfaction in immunodeficient mice infected with ZIKV. High viral load was observed in their olfactory system and downstream brain structures. ZIKV promoted both cellular damage in the olfactory neuroepithelium and protracted inflammation of the olfactory bulb, likely accounting for smell alteration. CONCLUSIONS: Patients with ZIKV-related GBS had poorer long-term olfactory function than patients with GBS-non-ZIKA, and ZIKV-infected mice are hyposmic. These observations suggest that ZIKV belongs on the list of viruses affecting the olfactory system. Clinical evaluation of the olfactory system should be considered for ZIKV-infected patients.


Asunto(s)
COVID-19 , Síndrome de Guillain-Barré , Infección por el Virus Zika , Virus Zika , Animales , Humanos , Ratones , Olfato , Infección por el Virus Zika/complicaciones , Infección por el Virus Zika/epidemiología
7.
Mamm Genome ; 32(1): 30-37, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33420513

RESUMEN

Rift Valley fever (RVF) is an emerging viral zoonosis that primarily affects ruminants and humans. We have previously shown that wild-derived MBT/Pas mice are highly susceptible to RVF virus and that part of this phenotype is controlled by a locus located on distal Chromosome 11. Using congenic strains, we narrowed down the critical interval to a 530 kb region containing five protein-coding genes among which Rnf213 emerged as a potential candidate. We generated Rnf213-deficient mice by CRISPR/CAS9 on the C57BL/6 J background and showed that they were significantly more susceptible to RVF than control mice, with an average survival time post-infection reduced from 7 to 4 days. The human RNF213 gene had been associated with the cerebrovascular Moyamoya disease (MMD or MYMY) but the inactivation of this gene in the mouse resulted only in mild anomalies of the neovascularization. This study provides the first evidence that the Rnf213 gene may also impact the resistance to infectious diseases such as RVF.


Asunto(s)
Adenosina Trifosfatasas/genética , Resistencia a la Enfermedad/genética , Interacciones Huésped-Patógeno/genética , Fiebre del Valle del Rift/genética , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/fisiología , Ubiquitina-Proteína Ligasas/genética , Animales , Sistemas CRISPR-Cas , Mapeo Cromosómico , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
8.
J Virol ; 94(3)2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31694939

RESUMEN

The explosive spread of Zika virus (ZIKV) has been associated with major variations in severe disease and congenital afflictions among infected populations, suggesting an influence of host genes. We investigated how genome-wide variants could impact susceptibility to ZIKV infection in mice. We first describe that the susceptibility of Ifnar1-knockout mice is largely influenced by their genetic background. We then show that Collaborative Cross (CC) mice, which exhibit a broad genetic diversity, in which the type I interferon receptor (IFNAR) was blocked by an anti-IFNAR antibody expressed phenotypes ranging from complete resistance to severe symptoms and death, with large variations in the peak and the rate of decrease in the plasma viral load, in the brain viral load, in brain histopathology, and in the viral replication rate in infected cells. The differences in susceptibility to ZIKV between CC strains correlated with the differences in susceptibility to dengue and West Nile viruses between the strains. We identified highly susceptible and resistant mouse strains as new models to investigate the mechanisms of human ZIKV disease and other flavivirus infections. Genetic analyses revealed that phenotypic variations are driven by multiple genes with small effects, reflecting the complexity of ZIKV disease susceptibility in the human population. Notably, our results rule out the possibility of a role of the Oas1b gene in the susceptibility to ZIKV. Altogether, the findings of this study emphasize the role of host genes in the pathogeny of ZIKV infection and lay the foundation for further genetic and mechanistic studies.IMPORTANCE In recent outbreaks, ZIKV has infected millions of people and induced rare but potentially severe complications, including Guillain-Barré syndrome and encephalitis in adults. While several viral sequence variants were proposed to enhance the pathogenicity of ZIKV, the influence of host genetic variants in mediating the clinical heterogeneity remains mostly unexplored. We addressed this question using a mouse panel which models the genetic diversity of the human population and a ZIKV strain from a recent clinical isolate. Through a combination of in vitro and in vivo approaches, we demonstrate that multiple host genetic variants determine viral replication in infected cells and the clinical severity, the kinetics of blood viral load, and brain pathology in mice. We describe new mouse models expressing high degrees of susceptibility or resistance to ZIKV and to other flaviviruses. These models will facilitate the identification and mechanistic characterization of host genes that influence ZIKV pathogenesis.


Asunto(s)
Encéfalo/virología , Ratones de Colaboración Cruzada/genética , Variación Genética , Replicación Viral/fisiología , Infección por el Virus Zika/virología , 2',5'-Oligoadenilato Sintetasa , Animales , Encéfalo/patología , Chlorocebus aethiops , Ratones de Colaboración Cruzada/virología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Receptor de Interferón alfa y beta , Células Vero , Carga Viral , Virus del Nilo Occidental , Virus Zika/inmunología , Infección por el Virus Zika/patología
9.
Genes Immun ; 21(6-8): 365-379, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33219336

RESUMEN

The innate immune response is the major front line of defense against viral infections. It involves hundreds of genes with antiviral properties which expression is induced by type I interferons (IFNs) and are therefore called interferon stimulated genes (ISGs). Type I IFNs are produced after viral recognition by pathogen recognition receptors, which trigger a cascade of activation events. Human and mouse studies have shown that defective type I IFNs induction may hamper the ability to control viral infections. In humans, moderate to high-effect variants have been identified in individuals with particularly severe complications following viral infection. In mice, functional studies using knock-out alleles have revealed the specific role of most genes of the IFN pathway. Here, we review the role of the molecular partners of the type I IFNs induction pathway and their implication in the control of viral infections and of their complications.


Asunto(s)
Predisposición Genética a la Enfermedad , Inmunidad Innata , Interferón Tipo I/genética , Virosis/genética , Animales , Humanos , Interferón Tipo I/metabolismo , Virosis/inmunología
10.
Mamm Genome ; 31(3-4): 69-76, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32124004

RESUMEN

Scientists from 12 countries met at the International Mammalian Genome Conference (IMGC) to share advances in mammalian genetics and genomics research. The event was held in Strasbourg, France and represents the city's second time hosting the IMGC. A diverse attendance of pre-doctoral and post-doctoral trainees, young investigators, established researchers, clinicians, bioinformaticians, and computational biologists enjoyed a rich scientific program of 63 oral presentations, 65 posters, and 5 workshops in the fields of epigenetics, system genetics, developmental biology, cancer, human disease modeling, technical advances, and bioinformatics. This report presents selected highlights of this meeting which illustrate how recent advances in mammalian genetic approaches have improved our ability to decipher complex biological mechanisms.


Asunto(s)
Genoma/genética , Mamíferos/genética , Animales , Biología Computacional/métodos , Epigenómica/métodos , Genómica/métodos , Humanos , Ratones Endogámicos C57BL
11.
Int J Mol Sci ; 21(22)2020 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-33198087

RESUMEN

Male fertility disorders often have their origin in disturbed spermatogenesis, which can be induced by genetic factors. In this study, we used interspecific recombinant congenic mouse strains (IRCS) to identify genes responsible for male infertility. Using ultrasonography, in vivo and in vitro fertilization (IVF) and electron microscopy, the phenotyping of several IRCS carrying mouse chromosome 1 segments of Mus spretus origin revealed a decrease in the ability of sperm to fertilize. This teratozoospermia included the abnormal anchoring of the acrosome to the nucleus and a persistence of residual bodies at the level of epididymal sperm midpiece. We identified a quantitative trait locus (QTL) responsible for these phenotypes and we have proposed a short list of candidate genes specifically expressed in spermatids. The future functional validation of candidate genes should allow the identification of new genes and mechanisms involved in male infertility.


Asunto(s)
Cromosomas Humanos Par 1/genética , Infertilidad Masculina/genética , Sitios de Carácter Cuantitativo/genética , Acrosoma/fisiología , Animales , Núcleo Celular/genética , Núcleo Celular/fisiología , Epidídimo/fisiología , Femenino , Humanos , Masculino , Ratones , Fenotipo , Espermátides/fisiología , Espermatogénesis/genética , Espermatozoides/fisiología , Teratozoospermia/genética
12.
Infect Immun ; 88(1)2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31636138

RESUMEN

Salmonella is an intracellular bacterium found in the gastrointestinal tract of mammalian, avian, and reptilian hosts. Mouse models have been extensively used to model in vivo distinct aspects of human Salmonella infections and have led to the identification of several host susceptibility genes. We have investigated the susceptibility of Collaborative Cross strains to intravenous infection with Salmonella enterica serovar Typhimurium as a model of human systemic invasive infection. In this model, strain CC042/GeniUnc (CC042) mice displayed extreme susceptibility with very high bacterial loads and mortality. CC042 mice showed lower spleen weights and decreased splenocyte numbers before and after infection, affecting mostly CD8+ T cells, B cells, and all myeloid cell populations, compared with control C57BL/6J mice. CC042 mice also had lower thymus weights with a reduced total number of thymocytes and double-negative and double-positive (CD4+, CD8+) thymocytes compared to C57BL/6J mice. Analysis of bone marrow-resident hematopoietic progenitors showed a strong bias against lymphoid-primed multipotent progenitors. An F2 cross between CC042 and C57BL/6N mice identified two loci on chromosome 7 (Stsl6 and Stsl7) associated with differences in bacterial loads. In the Stsl7 region, CC042 carried a loss-of-function variant, unique to this strain, in the integrin alpha L (Itgal) gene, the causative role of which was confirmed by a quantitative complementation test. Notably, Itgal loss of function increased the susceptibility to S. Typhimurium in a (C57BL/6J × CC042)F1 mouse background but not in a C57BL/6J mouse inbred background. These results further emphasize the utility of the Collaborative Cross to identify new host genetic variants controlling susceptibility to infections and improve our understanding of the function of the Itgal gene.


Asunto(s)
Bacteriemia/genética , Antígeno CD11a/deficiencia , Predisposición Genética a la Enfermedad , Mutación con Pérdida de Función , Infecciones por Salmonella/genética , Salmonella typhimurium/crecimiento & desarrollo , Animales , Bacteriemia/inmunología , Bacteriemia/patología , Carga Bacteriana , Médula Ósea/patología , Modelos Animales de Enfermedad , Genes , Subgrupos Linfocitarios/inmunología , Ratones , Ratones Endogámicos C57BL , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/patología , Serogrupo , Bazo/patología , Análisis de Supervivencia , Timo/patología
13.
BMC Genomics ; 19(1): 303, 2018 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-29703142

RESUMEN

BACKGROUND: Salmonella is a Gram-negative bacterium causing a wide range of clinical syndromes ranging from typhoid fever to diarrheic disease. Non-typhoidal Salmonella (NTS) serovars infect humans and animals, causing important health burden in the world. Susceptibility to salmonellosis varies between individuals under the control of host genes, as demonstrated by the identification of over 20 genetic loci in various mouse crosses. We have investigated the host response to S. Typhimurium infection in 35 Collaborative Cross (CC) strains, a genetic population which involves wild-derived strains that had not been previously assessed. RESULTS: One hundred and forty-eight mice from 35 CC strains were challenged intravenously with 1000 colony-forming units (CFUs) of S. Typhimurium. Bacterial load was measured in spleen and liver at day 4 post-infection. CC strains differed significantly (P < 0.0001) in spleen and liver bacterial loads, while sex and age had no effect. Two significant quantitative trait loci (QTLs) on chromosomes 8 and 10 and one suggestive QTL on chromosome 1 were found for spleen bacterial load, while two suggestive QTLs on chromosomes 6 and 17 were found for liver bacterial load. These QTLs are caused by distinct allelic patterns, principally involving alleles originating from the wild-derived founders. Using sequence variations between the eight CC founder strains combined with database mining for expression in target organs and known immune phenotypes, we were able to refine the QTLs intervals and establish a list of the most promising candidate genes. Furthermore, we identified one strain, CC042/GeniUnc (CC042), as highly susceptible to S. Typhimurium infection. CONCLUSIONS: By exploring a broader genetic variation, the Collaborative Cross population has revealed novel loci of resistance to Salmonella Typhimurium. It also led to the identification of CC042 as an extremely susceptible strain.


Asunto(s)
Cruzamientos Genéticos , Susceptibilidad a Enfermedades , Sitios de Carácter Cuantitativo , Salmonelosis Animal/genética , Salmonelosis Animal/microbiología , Salmonella typhimurium/fisiología , Animales , Mapeo Cromosómico , Femenino , Variación Genética , Genética de Población , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo
14.
Mamm Genome ; 29(7-8): 384-407, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30167843

RESUMEN

Flaviviruses are arthropod-borne viruses, several of which represent emerging or re-emerging pathogens responsible for widespread infections with consequences ranging from asymptomatic seroconversion to severe clinical diseases and congenital developmental deficits. This variability is due to multiple factors including host genetic determinants, the role of which has been investigated in mouse models and human genetic studies. In this review, we provide an overview of the host genes and variants which modify susceptibility or resistance to major mosquito-borne flaviviruses infections in mice and humans.


Asunto(s)
Culicidae/virología , Infecciones por Flavivirus/genética , Infecciones por Flavivirus/virología , Flavivirus/fisiología , Predisposición Genética a la Enfermedad , Interacciones Huésped-Patógeno/genética , Animales , Biomarcadores , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Infecciones por Flavivirus/inmunología , Infecciones por Flavivirus/transmisión , Estudio de Asociación del Genoma Completo , Interacciones Huésped-Patógeno/inmunología , Humanos , Ratones
15.
Mamm Genome ; 29(7-8): 558-576, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29785663

RESUMEN

Enterobacteriaceae are a large family of Gram-negative, non-spore-forming bacteria. Although many species exist as part of the natural flora of animals including humans, some members are associated with both intestinal and extraintestinal diseases. In this review, we focus on members of this family that have important roles in human disease: Salmonella, Escherichia, Shigella, and Yersinia, providing a brief overview of the disease caused by these bacteria, highlighting the contribution of animal models to our understanding of their pathogenesis and of host genetic determinants involved in susceptibility or resistance to infection.


Asunto(s)
Resistencia a la Enfermedad , Susceptibilidad a Enfermedades , Infecciones por Enterobacteriaceae/microbiología , Enterobacteriaceae/fisiología , Interacciones Huésped-Patógeno , Alelos , Animales , Modelos Animales de Enfermedad , Resistencia a la Enfermedad/genética , Resistencia a la Enfermedad/inmunología , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/metabolismo , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Sitios de Carácter Cuantitativo
16.
J Infect Dis ; 216(6): 761-770, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28934426

RESUMEN

Background: Susceptibility to infection is in part genetically driven, and C57BL/6 mice resist various pathogens through the proinflammatory response of their M1 macrophages (MPs). However, they are susceptible to plague. It has been reported elsewhere that Mus spretus SEG mice resist plague and develop an immune response characterized by a strong recruitment of MPs. Methods: The responses of C57BL/6 and SEG MPs exposed to Yersinia pestis in vitro were examined. Results: SEG MPs exhibit a stronger bactericidal activity with higher nitric oxide production, a more proinflammatory polarized cytokine response, and a higher resistance to Y. pestis-induced apoptosis. This response was not specific to Y. pestis and involved a reduced sensitivity to M2 polarization/signal transducer and activator of transcription 6 activation and inhibition of caspase 8. The enhanced M1 profile was inducible in C57BL/6 MPs in vitro, and when transferred to susceptible C57BL/6 mice, these MPs significantly increased survival of bubonic plague. Conclusions: MPs can develop an enhanced functional profile beyond the prototypic M1, characterized by an even more potent proinflammatory response coordinated with resistance to killing. This programming plays a key role in the plague-resistance phenotype and may be similarly significant in other highly lethal infections, suggesting that orienting the MP response may represent a new therapeutic approach.


Asunto(s)
Apoptosis , Macrófagos/inmunología , Peste/inmunología , Yersinia pestis , Animales , Células Cultivadas , Citocinas/sangre , Femenino , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Óxido Nítrico/metabolismo , Transducción de Señal
17.
Int J Cancer ; 139(6): 1358-71, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27130719

RESUMEN

Enhancing anti-tumor immunity and preventing tumor escape are efficient strategies to increase the efficacy of therapeutic cancer vaccines. However, the treatment of advanced tumors remains difficult, mainly due to the immunosuppressive tumor microenvironment. Regulatory T cells and myeloid-derived suppressor cells have been extensively studied, and their role in suppressing tumor immunity is now well established. In contrast, the role of B lymphocytes in tumor immunity remains unclear because B cells can promote tumor immunity or display regulatory functions to control excessive inflammation, mainly through IL-10 secretion. Here, in a mouse model of HPV-related cancer, we demonstrate that B cells accumulated in the draining lymph node of tumor-bearing mice, due to a prolonged survival, and showed a decreased expression of MHC class II and CD86 molecules and an increased expression of Ly6A/E, PD-L1 and CD39, suggesting potential immunoregulatory properties. However, B cells from tumor-bearing mice did not show an increased ability to secrete IL-10 and a deficiency in IL-10 production did not impair tumor growth. In contrast, in B cell-deficient µMT mice, tumor rejection occurred due to a strong T cell-dependent anti-tumor response. Genetic analysis based on single nucleotide polymorphisms identified genetic variants associated with tumor rejection in µMT mice, which could potentially affect reactive oxygen species production and NK cell activity. Our results demonstrate that B cells play a detrimental role in anti-tumor immunity and suggest that targeting B cells could enhance the anti-tumor response and improve the efficacy of therapeutic cancer vaccines.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Infecciones por Papillomavirus/complicaciones , Neoplasias del Cuello Uterino/etiología , Neoplasias del Cuello Uterino/metabolismo , Animales , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Subgrupos de Linfocitos B/patología , Linfocitos B/patología , Movimiento Celular/inmunología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Estudio de Asociación del Genoma Completo , Interleucina-10/biosíntesis , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Activación de Linfocitos , Ratones , Papillomaviridae , Infecciones por Papillomavirus/virología , Fenotipo , Polimorfismo de Nucleótido Simple , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Receptor Toll-Like 9/metabolismo , Neoplasias del Cuello Uterino/patología
18.
Am J Pathol ; 184(2): 362-8, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24296104

RESUMEN

Fertility is a quantitative, complex character governed by a considerable number of genes. Despite clinical and scientific advances, several cases of human infertility remain unexplained. In the present study, using a positional cloning approach in a mouse model of interspecific recombinant lines, a candidate gene, ALPP, encoding the placental alkaline phosphatase, was identified as being potentially involved in recurrent spontaneous abortion. We then analyzed patients for detecting putative associations between ALPP polymorphisms, in vitro fertilization failures, and miscarriages. ALPP was sequenced in 100 controls and 100 patients affected by recurrent spontaneous abortion, from the same ethnic background. The frequency of several alleles and allelic combinations were different between recurrent spontaneous abortion and control women. One polymorphism induced a coding substitution (Ile89Leu) that was associated with a decreased risk of abortion and in vitro fertilization failure. Thereafter, the population was increased by the analysis of 92 additional controls and 612 additional patients for the coding polymorphism Ile89Leu. We finally show, by functional analysis, that the 89Leu placental alkaline phosphatase has an enhanced alkaline phosphatase activity. This study suggests that ALPP genotyping could be a strong predictor of implantation success.


Asunto(s)
Aborto Espontáneo/enzimología , Aborto Espontáneo/genética , Fosfatasa Alcalina/genética , Fertilización In Vitro , Predisposición Genética a la Enfermedad , Isoenzimas/genética , Polimorfismo de Nucleótido Simple/genética , Animales , Células COS , Chlorocebus aethiops , Estudios de Cohortes , Femenino , Proteínas Ligadas a GPI/genética , Técnicas de Genotipaje , Humanos , Ratones , Embarazo , Recurrencia , Reproducibilidad de los Resultados , Transfección
19.
J Exp Med ; 221(2)2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38117256

RESUMEN

Mucosal-associated invariant T (MAIT) cells harbor evolutionarily conserved TCRs, suggesting important functions. As human and mouse MAIT functional programs appear distinct, the evolutionarily conserved MAIT functional features remain unidentified. Using species-specific tetramers coupled to single-cell RNA sequencing, we characterized MAIT cell development in six species spanning 110 million years of evolution. Cross-species analyses revealed conserved transcriptional events underlying MAIT cell maturation, marked by ZBTB16 induction in all species. MAIT cells in human, sheep, cattle, and opossum acquired a shared type-1/17 transcriptional program, reflecting ancestral features. This program was also acquired by human iNKT cells, indicating common differentiation for innate-like T cells. Distinct type-1 and type-17 MAIT subsets developed in rodents, including pet mice and genetically diverse mouse strains. However, MAIT cells further matured in mouse intestines to acquire a remarkably conserved program characterized by concomitant expression of type-1, type-17, cytotoxicity, and tissue-repair genes. Altogether, the study provides a unifying view of the transcriptional features of innate-like T cells across evolution.


Asunto(s)
Células T Invariantes Asociadas a Mucosa , Humanos , Bovinos , Animales , Ratones , Ovinos , Diferenciación Celular , Membrana Celular , Reparación por Escisión , Especificidad de la Especie , Mamíferos/genética
20.
J Infect Dis ; 205(1): 134-43, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22090450

RESUMEN

BACKGROUND: Although laboratory mice are usually highly susceptible to Yersinia pestis, we recently identified a mouse strain (SEG) that exhibited an exceptional capacity to resist bubonic plague and used it to identify immune mechanisms associated with resistance. METHODS: The kinetics of infection, circulating blood cells, granulopoiesis, lesions, and cellular populations in the spleen, and cytokine production in various tissues were compared in SEG and susceptible C57BL/6J mice after subcutaneous infection with the virulent Y. pestis CO92. RESULTS: Bacterial invasion occurred early (day 2) but was transient in SEG/Pas mice, whereas in C57BL/6J mice it was delayed but continuous until death. The bacterial load in all organs significantly correlated with the production of 5 cytokines (granulocyte colony-stimulating factor, keratinocyte-derived chemokine (KC), macrophage cationic peptide-1 (MCP-1), interleukin 1α, and interleukin 6) involved in monocyte and neutrophil recruitment. Indeed, higher proportions of these 2 cell types in blood and massive recruitment of F4/80(+)CD11b(-) macrophages in the spleen were observed in SEG/Pas mice at an early time point (day 2). Later times after infection (day 4) were characterized in C57BL/6J mice by destructive lesions of the spleen and impaired granulopoiesis. CONCLUSION: A fast and efficient Y. pestis dissemination in SEG mice may be critical for the triggering of an early and effective innate immune response necessary for surviving plague.


Asunto(s)
Citocinas/metabolismo , Inmunidad Innata , Ratones Endogámicos/inmunología , Peste/inmunología , Yersinia pestis/patogenicidad , Animales , Carga Bacteriana , Quimiocinas/metabolismo , Resistencia a la Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos/metabolismo , Fagocitos/inmunología , Peste/metabolismo , Peste/microbiología , Yersinia pestis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA